throbber
Journal of Steroid Biochemistry & Molecular Biology 84 (2003) 537–542
`
`In vitro and in vivo models for the evaluation of potent inhibitors
`of male rat 17␣-hydroxylase/C17,20-lyase
`I. Duc a,∗
`, P. Bonnet a, V. Duranti a, S. Cardinali a, A. Rivière a, A. De Giovanni a,
`J. Shields-Botella a, G. Barcelo b, N. Adje b, D. Carniato b, J. Lafay b,
`J.C. Pascal b, R. Delansorne a
`a Preclinical R&D Department, Théramex, 6 Avenue Prince Héréditaire, Albert 98000, Monaco
`b Chemical R&D Department, Théramex, 6 Avenue Prince Héréditaire, Albert 98000, Monaco
`
`Received 19 August 2002; accepted 27 January 2003
`
`Abstract
`
`The C17,20-lyase is a key enzyme in the biosynthesis of androgens by both the testes and adrenals. A complete inhibition of this enzyme
`would provide an alternative means of androgen suppression for the treatment of prostatic cancers. In the present study, the inhibitory
`effects of new non-steroidal compounds were tested in vitro on rat C17,20-lyase versus abiraterone, a reference steroidal inhibitor. Their
`activities were also evaluated in vivo on plasma testosterone (T) and luteinizing hormone (LH) levels and on testes, adrenals, seminal
`vesicles (SV) and ventral prostate (VP) weights after 3 days of oral treatment to adult male rats (50 mg/kg per day p.o.).
`Inhibition in the nanomolar range was obtained with TX 977, the lead racemate product in this series, and optimization is ongoing based
`on a slight dissociation observed between its two diastereoisomers, TX 1196-11 (S) and TX 1197-11 (R). These non-steroidal compounds
`(including YM 55208, a reference competitor) proved to be more active in vivo than abiraterone acetate in this model, but the observed
`impact on adrenal weight suggests that the specificity of lyase inhibition versus corticosteroid biosynthesis deserves further investigations
`with this new class of potentially useful agents for the treatment of androgen-dependent prostate cancer.
`© 2003 Elsevier Science Ltd. All rights reserved.
`
`Keywords: Steroidogenesis; Androgen; P450C17; Inhibitor of C17,20-lyase; Rat
`
`1. Introduction
`
`The 17␣-hydroxylase/C17,20-lyase plays a key part in
`the pathways of steroid hormone biosynthesis [1,2]. This
`enzyme catalyses two reactions: 17␣ hydroxylation of C21
`steroids and cleavage of C17,20 bond of C21 steroids. The
`17␣ hydroxylation activity is a required step in cortisol
`biosynthesis whereas the C17,20 bond side chain cleavage
`is essential for the biosynthesis of androgens. The 17␣-
`hydroxylase/C17,20-lyase is a cytochrome P450-dependent
`microsomal enzyme (P450C17), which is expressed in tes-
`ticular and adrenal tissues and catalyses the conversion of
`pregnenolone or progesterone into dehydroepiandrosterone
`(DHEA) or androstenedione ( 4A), respectively, two pre-
`cursors of testosterone (T) [3–7]. The use of effective
`and selective inhibitor of P450C17 appears as a possible
`alternative to orchidectomy or other endocrine therapy,
`
`Corresponding author. Tel.: +377-92-05-08-58;
`∗
`fax: +377-92-05-08-81.
`E-mail address: iduc@theramex.mc (I. Duc).
`
`to lower circulating androgens in patients with prostate
`cancer.
`In the rat and other rodents, 17␣-hydroxylase/C17,20-lyase
`is not expressed in zona fasciculata of adrenal cortex, con-
`sequently corticosterone and not cortisol as in human, is
`the major glucocorticoid. Moreover, the rat P450C17 can
`convert both 17␣-hydroxy-pregnenolone and 17␣-hydroxy-
`progesterone (17OHP) into DHEA and 4A, respectively,
`as compared to human and bovine enzyme that can only
`cleave the C17,20 bond of 17␣-hydroxy-pregnenolone [8,9].
`Even so, due to the lack of availability of human tissue and
`variability among samples, the rat remains, in spite of dif-
`ferences in localization and substrate specificity of this en-
`zyme, a suitable model for the in vitro and in vivo evaluation
`of inhibitory potential of new compounds [10,11].
`The first aim of this study was, to compare in vitro, the
`inhibitory activity of new steroidal and non-steroidal com-
`pounds on C17,20-lyase from rat testis microsomes. 17OHP
`was chosen as substrate, and the inhibitory potential of test
`compounds was determined by measuring the end product
`of the reaction, 4A, by radioimmunoassay (RIA).
`
`0960-0760/03/$ – see front matter © 2003 Elsevier Science Ltd. All rights reserved.
`doi:10.1016/S0960-0760(03)00078-5
`
`JANSSEN EXHIBIT 2012
`Mylan v. Janssen IPR2016-01332
`
`

`

`I. Duc et al. / Journal of Steroid Biochemistry & Molecular Biology 84 (2003) 537–542
`tions for storage at −70
`◦
`C. The protein concentration of the
`suspension was determined by the method of Bradford [21]
`with the Bio-Rad protein assay kit.
`
`538
`
`the inhibitory potential of the compounds
`Secondly,
`was evaluated in vivo in adult male rats after 3 days of
`oral treatment. This in vivo model allowed a rapid screen-
`ing of new compounds as compared to the classic 15-
`day model [12]. The weights of androgen-target organs
`such as ventral prostate (VP) and seminal vesicles (SV)
`were measured as well as circulating levels of T and LH.
`The weights of androgen-sensitive organs such as testes
`and adrenal glands were also monitored as an index of
`side-effects. Abiraterone (CB7598), abiraterone acetate
`(CB7630), steroid derivatives [13] and YM 55208 [14], a
`non-steroid compound derived from YM 116 [15], were
`used as references as well as the anti-fungal bifonazole
`[16–18].
`
`2.4. C17,20-lyase activity assay
`
`Microsomes were diluted to a final protein concentra-
`tion of 50 ␮g/ml in the reaction mixture which contained
`0.25 M sucrose, 20 mM Tris–HCl (pH 7.4), 10 mM G6P
`◦
`and 1.2 IU/ml G6PDH. After equilibration at 37
`C for
`10 min, the reaction was initiated by addition of ␤NADP
`to obtain a final concentration of 0.6 mM. Prior to the
`distribution of 600 ␮l of the reaction mixture in each
`tube, test compounds were evaporated to dryness under a
`◦
`stream of nitrogen and then were incubated at 37
`C for
`10 min.
`After incubation with inhibitors, 500 ␮l of the reaction
`mixture was transferred to tubes containing 1 ␮M of the
`enzyme substrate, 17OHP. After a further 10 min incubation,
`tubes were placed on ice and the reaction was stopped by
`addition of 0.1 ml NaOH 1N. Tubes were deep-frozen and
`stored at −20
`◦
`C until assayed for 4A levels.
`A 4A RIA was developed and automated on a mi-
`croplate format in our laboratory using a specific antibody
`against 4A and instructions provided by Biogenesis
`(Poole, England). The separation of free and bound antigen
`was achieved with a dextran-coated charcoal suspension.
`After centrifugation, aliquots of the clear supernatant were
`counted in duplicates in a 1450 MicrobetaPlus liquid scin-
`tillation counter (Perkin-Elmer Instruments, Courtaboeuf,
`France). The 4A concentrations of unknown samples
`were determined from the standard curve. The detection
`limit was 0.5 ng/ml and the within and between assay coef-
`ficients of variation were 10.7 and 17.6%, respectively at an
`assay value of 13 ng/ml. The rate of enzymatic reaction was
`expressed as pmol of 4A formed per 10 min and per mg
`of protein. The value of maximum activity without inhibitor
`(control) was set at 100%. The IC50 values were calculated
`using non-linear analysis from the plot of enzyme activity
`(%) against log of inhibitor concentration (GraphPad Prism,
`version 3.0).
`
`2.5. In vivo assays
`
`Adult male Wistar rats weighing 220–240 g were used.
`All the tested compounds were prepared in distilled water
`with a few drops of Tween 80 (polyoxyethylene sorbitan
`monoleate) and administered by oral route at 10 ml/kg per
`day to animals once daily for 3 days. Control group was
`dosed with vehicle (water-Tween 80) and test compounds
`were given orally at 50 mg/kg per day. On the day following
`the last treatment, animals were anaesthetized with isoflu-
`rane. VP, SV, testes and adrenals were removed, dissected
`and weighed. Arterial blood was withdrawn and collected
`into heparinized tubes. Plasma was stored at −20
`◦
`C until
`required.
`
`2. Materials and methods
`
`2.1. Chemical inhibitors and radioactive steroids
`
`TX 977 (racemate), TX 977-11 (salt of TX 977),
`TX 1196-11 (S-enantiomer of TX 977), TX 1197-11
`(R-enantiomer of TX 977), abiraterone (3␤-hydroxy-
`17-(3-pyridyl)-androsta-5,16-diene),
`abiraterone
`acetate
`(3␤-acetoxy-17-(3-pyridyl)-androsta-5,16-diene), and YM
`55208 (2-(1-(1H-imidazol-1-yl)ethyl)-9H-carbazole) were
`synthesized by Théramex (Monaco). Bifonazole (1-(p,␣-
`diphenylbenzyl)-imidazole) was purchased from Sigma
`(St. Louis, MO, USA). [1,2,6,7-3H]-Androt-4-ene-3,17-
`dione(3H- 4A) was purchased from NEN Life Science
`Products (Boston, MA, USA).
`Other not listed or not specified compounds and reagents
`were from Sigma (St. Quentin Fallavier, France).
`
`2.2. Animals
`
`Adult Wistar male rats were selected for the present study.
`They were purchased from Iffa Credo (Elevage des Oncins,
`L’Arbresle, France). They were housed under conditions of
`12 h light–dark, maintained in an air-conditioned room and
`provided with a standard diet of AO4C pellets from UAR
`(Villemoisson-sur-Orge, France) and filtered mains water ad
`libitum.
`
`2.3. Preparation of rat testis microsomes
`
`Testes from 230 to 310 g rats were obtained by scrotal
`castration. They were washed with an isotonic solution of
`NaCl, pooled, weighed and then blended with an ultra-turrax
`homogenizer in 0.25 M sucrose, 20 mM Tris–HCl (pH 7.4).
`The homogenate was centrifuged at 10 000× g for 30 min at
`C. The supernatant obtained was centrifuged at 100 000×
`◦
`4
`◦
`g for 60 min at 4
`C. The microsomal pellet [19,20] thus ob-
`tained was resuspended in 5 mM MgCl2, 50 mM Tris–HCl,
`pH 7.4 buffer, treated briefly with a dual/kontes homoge-
`nizer to ensure full dispersion and divided into 400 ␮l por-
`
`

`

`I. Duc et al. / Journal of Steroid Biochemistry & Molecular Biology 84 (2003) 537–542
`
`539
`
`2.6. T and LH RIA assays
`
`Plasma was used for the determination of T as described
`in the 125I-T assay kit supplied by Diagnostic Systems Lab-
`oratories Inc. (Webster, TX, USA). The detection limit was
`0.1 ng/ml and the within and between assay coefficients of
`variation were 13.9 and 13.7%, respectively at an assay value
`of 0.5 ng/ml.
`The assay for LH used the rat specific 125I-LH assay
`system from Amersham Biosciences Europe GmbH (Orsay,
`France). The detection limit was 0.8 ng/ml and the within
`and between assay coefficients of variation were 5.2 and
`1.9%, respectively at an assay value of 3.1 ng/ml.
`
`2.7. Statistical analysis
`
`Statistical analysis was performed using the SAS soft-
`ware version 6.12 (SAS Institute, Grégy-Sur-Yerres, FR).
`For IC50, organ weights and plasma hormone levels, homo-
`geneity of variances was checked by Levene’s test. Results
`were analyzed using the parametric analysis of variance
`followed by multiple range tests or the non-parametric
`Kruskall–Wallis analysis and Wilcoxon tests, depending on
`the homogeneity of the variances. A P value less than 0.05
`was considered as statistically significant.
`
`3. Results
`
`3.1. Inhibition of rat testicular C17,20-lyase
`
`As shown in Fig. 1, a concentration-related inhibition
`of the rat testicular C17,20-lyase activity was obtained with
`
`Fig. 1. Inhibition of rat testicular C17,20-lyase activity. Rat testes micro-
`somes were prepared as described in Section 2. C17,20-lyase activity was
`measured after incubation with 1 ␮M of 17OHP and the indicated con-
`centrations of compounds. Each point represents the mean ± S.E.M. of
`four determinations each made in duplicate in individual experiments.
`
`Table 1
`IC50 for the inhibition of lyase activity in rat testis microsomes
`
`Compound
`
`IC50 (nM)
`5.8 ± 0.8a
`Abiraterone
`6.2 ± 0.5
`YM 55208
`6.4 ± 1.0a
`TX 1196-11, S-enantiomer
`8.7 ± 2.0a
`TX 977 racemate
`13.6 ± 3.4
`∗
`TX 1197-11, R-enantiomer
`21.5 ± 5.6
`∗
`Bifonazole
`Means ± S.E.M. for four determinations.
`a NS: P > 0.05.
`∗
`0.01 < P < 0.05 as compared with YM 55208.
`
`all the test compounds. The residual activity expressed as
`the percentage of total activity obtained in presence of test
`compounds, decreased within a range of 115–1% from 1 to
`128 nM. All test compounds were shown to be potent in-
`hibitors of the C17,20-lyase with IC50 values in the nanomo-
`lar range as summarized in Table 1. The decreasing sequence
`of the inhibitory activity was as follows:
`abiraterone ≥ YM 55208 ≥ TX 1196-11(S-enantiomer)
`≥ TX 977(racemate)
`≥ TX 1197-11(R-enantiomer) ≥ Bifonazole.
`The racemate product and its S-enantiomer, were as po-
`tent as abiraterone and YM 55208,
`the steroidal and
`non-steroidal references, respectively. There was a slight
`but not statistically significant difference between the in-
`hibitory potencies of the two diastereoisomers. Bifonazole
`and the R-enantiomer were the less potent compounds in
`this in vitro model.
`
`3.2. Organ weights
`
`As shown in Fig. 2, after 3 days of oral treatment at
`50 mg/kg per day, abiraterone acetate, markedly inhibited
`VP (−14%) and SV weights (−37%) without affecting
`adrenal weight (−7%). YM 55208 induced a more potent
`reduction in VP (−37%) and SV (−48%) but exhibited an
`harmful effect on adrenals by increasing their weight by
`17%. The racemate compound and its S-enantiomer, pro-
`duced similar notable reductions in VP (−24 and −22%,
`respectively) and SV (−42 and −41%, respectively) but the
`S-enantiomer only caused a slight but statistically significant
`increase in adrenal weight (+11%).
`The R-enantiomer caused a marked reduction in VP
`(−24%) which was comparable to that obtained with its two
`related compounds. However, it induced a weaker inhibi-
`tion of SV (−22%), while the increase observed in adrenal
`weight (15%) was not statistically significant. None of the
`tested compounds modified testis weight.
`
`3.3. Plasma hormone levels
`
`Fig. 2 shows that when administered orally at 50 mg/kg
`per day to adult male rat, abiraterone acetate significantly
`
`

`

`540
`
`I. Duc et al. / Journal of Steroid Biochemistry & Molecular Biology 84 (2003) 537–542
`
`rise in LH secretion (378%). The racemate product and its
`S-enantiomer induced a 89% inhibition on T secretion and
`increased LH secretion by 240 and 225%, respectively. The
`R-enantiomer significantly inhibited T levels (−63%) and
`increased LH levels (225%) but its effect on plasma T ap-
`peared to be less marked than that of its related compounds.
`
`4. Discussion
`
`In the search of potent inhibitors of C17,20-lyase, the key
`enzyme in androgen biosynthesis, a chemical series of new
`non-steroidal inhibitors was synthesized and tested in vitro
`and in vivo on the rat C17,20-lyase.
`In the in vitro assay reported here, the HPLC product iso-
`lation [22–25] or the classical measurement of the 3H-acetic
`acid released [26–28] were substituted with a RIA of 4A
`produced from the substrate 17OHP. Optimum conditions
`for rat testicular microsome C17,20-lyase activity measure-
`ment with respect to substrate, cofactor, and protein concen-
`trations, as well as time and compatibility with the range of
` 4A in the RIA were determined during preliminary exper-
`iments, and are described in details in Section 2. The activ-
`ity of the control reaction (substrate without enzyme) was
`found to be less than 4% of the enzyme activity. The kinetic
`study of the C17,20-lyase in our conditions indicated that the
`Km was 230 nM and the Vmax was 260 pmol/(min mg) of
`protein (results not shown).
`With this in vitro assay, a chemical series of new
`non-steroidal compounds was investigated for its inhibitory
`potency on rat C17,20-lyase activity (results not shown).
`One of the compounds was isolated as a lead, TX 977 a
`racemate product, and its salt TX 977-11 was used in in
`vivo assays. Its two diastereoisomers, TX 1196-11 (S) and
`TX 1197-11 (R), were then synthesized and evaluated in
`comparison with abiraterone [29] and YM 55208 [14], as
`a steroidal and a non-steroidal references, respectively, and
`the anti-fungal bifonazole [16]. In vitro, a slight difference
`was observed between the two enantiomers, in favor of the
`S-compound. However, the three compounds were as potent
`as abiraterone and YM 55208 with IC50 in the nanomolar
`range, while bifonazole was the less active compound.
`In order to obtain a fast evaluation of the in vivo activ-
`ity of our new compounds, a short model based on a 3-day
`treatment by oral route and a sacrifice 24 h after the last ad-
`ministration was set-up. In this model, the known inhibitory
`effects of abiraterone acetate [30] and YM 55208 [12] on
`VP and SV weights were reproduced. They were directly
`related to the decrease in circulating T levels obtained by
`inhibition of the C17,20-lyase activity. This decrease in T
`levels was associated with a rise in plasma LH, illustrating
`the absence of the negative feedback of T on the pituitary
`gland. Despite this LH increase, which should stimulate the
`testicular androgen production, YM 55208 was able to main-
`tain very low levels of T, below the LOQ of the assay that
`is 0.1 ng/ml. The lead compound and its two enantiomers
`
`Fig. 2. Anti-androgenic activity in the rat. Adult rats were treated as
`described in Section 2. They were treated orally with 50 mg/kg per day
`for 3 days. On the day following the last treatment, ventral prostate
`seminal vesicles (A), testes and adrenal glands (B) were removed and
`weighed. Arterial blood was collected and stored at −20
`◦
`C till T and
`LH assays (C). Each bar represents the mean ± S.E.M. of at least eight
`∗P < 0.05,
`∗∗P < 0.01,
`∗∗∗P < 0.005 as compared with
`determinations.
`control group.
`
`inhibited T secretion (−48%) and in turn increased LH con-
`centration (192%). YM 55208 was shown to totally inhibit
`plasma T secretion (more than −97%) with levels lower
`than the LOQ. This inhibitory effect, more potent than that
`of abiraterone acetate was also associated with a feedback
`
`

`

`I. Duc et al. / Journal of Steroid Biochemistry & Molecular Biology 84 (2003) 537–542
`
`541
`
`were shown to be more potent than abiraterone acetate on
`VP weights and T levels. As in vitro, a slight dissociation
`in favor of the S- versus the R-enantiomer was observed for
`SV weights and T levels. No side-effects were observed for
`none of the test compounds on testis (Fig. 2), liver or body
`weights (results not shown).
`However, the S-enantiomer was found to induce an in-
`crease on adrenal weight. This side-effect although weaker
`than that observed with YM 55208 may indicate a lack of
`specificity in inhibition. Then, since in rat the biosynthesis
`of corticosterone does not require the cytochrome P450C17
`enzyme, a selective inhibitor should not have an effect on
`adrenal function and particularly no effect on corticosterone
`levels. In an attempt to explore this effect on adrenals, cor-
`ticosterone levels were measured after a 1-week treatment
`with the S-enantiomer at the dose of 10 mg/kg per day (re-
`sult not shown). There was no statistically significant dif-
`ference observed in corticosterone levels as compared with
`the intact control group, indicating that this new compound
`is probably devoid of inhibitory activity on other enzymes
`involved in corticosterone biosynthesis.
`In conclusion, optimization is ongoing based on the
`nanomolar range inhibition obtained with TX 977-11 the
`lead of our new chemical series of non-steroidal com-
`pounds and the slight dissociation observed between its two
`diastereoisomers, TX 1196-11 (S) and TX 1197-11 (R).
`Together with YM 55208 in our model, they proved to be
`more active in vivo than abiraterone acetate. However, the
`impact observed on adrenal weight suggests that the speci-
`ficity of lyase inhibition versus corticosteroid biosynthesis
`deserve further investigations with this new class of poten-
`tially useful agents for the treatment of androgen-dependent
`prostate cancer. Specificity, pharmacokinetic and long-term
`models (4 weeks at least) are now in progress.
`
`References
`
`[1] W.L. Miller, Molecular biology of steroid hormone synthesis,
`Endocrinol. Rev. 9 (1988) 295–318.
`[2] I. Hanukoglu, Steroidogenic enzymes: structure, function, and role
`in regulation of steroid hormone biosynthesis, J. Steroid Biochem.
`Mol. Biol. 43 (8) (1992) 779–804.
`steroid
`of P450-dependent
`[3] H. Vanden Bossche,
`Inhibitors
`biosynthesis: from research to medical treatment, J. Steroid Biochem.
`Mol. Biol. 43 (8) (1992) 1003–1021.
`of
`[4] D.F.V. Lewis, P. Lee-Robichaud, Molecular modelling
`steroidogenic cytochromes P450 from families CYP11, CYP17,
`CYP19 and CYP21 based on the CYP102 crystal structure, J. Steroid
`Biochem. Mol. Biol. 66 (4) (1998) 217–233.
`[5] W.L. Miller, R.J. Auchus, D.H. Geller, The regulation of 17,20 lyase
`activity, Steroids 62 (1997) 133–142.
`testis
`[6] M. Namiki, M. Kitamura, E. Buczko, M.L. Dufau, Rat
`P-450(17)alpha cDNA: the deduced amino acid sequence, Biochem.
`Biophys. Res. Commun. 157 (2) (1988) 705–712.
`[7] R.J. Auchus, W.L. Miller, Molecular modeling of human P450c17
`(17␣-hydroxylase/17,20-lyase): Insights into reaction mechanisms
`and effects of mutations, Mol. Endocrinol. 13 (7) (1999) 1169–1182.
`I.M. Bird, The role of cytochrome P450 17␣-
`[8] A.J. Conley,
`hydroxylase and 3␤-hydroxysteroid dehydrogenase in the integration
`
`of gonadal and adrenal steroidogenesis via the 5 and 4 pathways
`of steroidogenesis in mammals, Biol. Reprod. 56 (1997) 789–799.
`[9] B.J. Brock, M.R. Waterman, Biochemical differences between rat
`and human cytochrome P450c17 support the different steroidogenic
`needs of these two species, Biochemistry 38 (5) (1999) 1598–1606.
`[10] R.W. Hartmann, M. Hector, B.G. Wachall, A. Palusczak, M. Palzer, V.
`Huch, M. Veith, Synthesis and evaluation of 17-aliphatic heterocycle-
`substituted steroidal inhibitors of 17alpha-hydroxylase/C17-20-lyase
`(P450 17), J. Med. Chem. 43 (23) (2000) 4437–4445.
`[11] I.P. Nnane, K. Kato, Y. Liu, B.J. Long, Q. Lu, X. Wang, Y.-Z. Ling,
`A. Brodie, Inhibition of androgen synthesis in human testicular and
`prostatic microsomes and in male rats by novel steroidal compounds,
`Endocrinology 140 (6) (1999) 2891–2897.
`[12] J. Li, Y. Li, C. Son, P. Banks, A. Brodie, 4-Pregnene-3-one-20␤-
`carboxaldehyde: a potent inhibitor of 17␣-hydroxylase/C17,20-lyase
`and of 5␣-reductase, J. Steroid Biochem. Mol. Biol. 42 (3/4) (1992)
`313–320.
`[13] S.E. Barrie, M. Jarman, G.A. Potter, British Technology Group
`Limited, UK, 17-Substituted steroids useful in cancer treatment, UK
`Patent Applic. 2,265,624 (1993).
`[14] M. Okada, T. Yoden, E. Kawaminami, Y. Shimada, T. Ishihara,
`M. Kudoh, Yamanoushi Pharmaceutical Co. Ltd., Japan. Preparation
`of azole derivatives as steroid 17–20 lyase inhibitors, WO Patent
`9,509,157 (1994).
`[15] Y. Ideyama, M. Kudoh, K. Tanimoto, Y. Susaki, T. Nanya, T.
`Nakahara, H. Ishikawa, T. Yoden, M. Okada, T. Fujikura, H. Akaza,
`H. Shikama, Novel nonsteroidal inhibitor of cytochrome P45017␣
`(17␣-hydroxylase/C17-20 lyase), YM 116, decreased prostatic
`weights by reducing serum concentrations of T and adrenal androgens
`in rats, Prostate 37 (1998) 10–18.
`[16] M. Ayub, M.J. Levell, Inhibition of testicular 17␣-hydroxylase
`and 17,20-lyase but not 3␤-hydroxysteroid dehydrogenase-isomerase
`or 17␤-hydroxysteroid oxidoreductase by ketoconazole and other
`imidazole drugs, J. Steroid Biochem. 28 (5) (1987) 521–531.
`[17] M. Ayub, M.J. Levell, Inhibition of human adrenal steroidogenic
`enzymes in vitro by imidazole drugs including ketoconazole, J.
`Steroid Biochem. 32 (4) (1989) 515–524.
`[18] H. Vanden Bossche, P. Marichal, J. Gorrens, M.-C. Coene, G.
`Willemsens, D. Bellens, I. Roels, H. Moereels, P.A.J. Janssen,
`Biochemical approaches to selective antifungal activity. Focus on
`azole antifungals, Mycoses 32 (Suppl. 1) (1989) 35–52.
`[19] P.B. Kan, M.A. Hirst, D. Feldman, Inhibition of steroidogenic
`cytochrome P-450 enzymes in rat testis by ketoconazole and related
`imidazole anti-fungal drugs, J. Steroid Biochem. 23 (6A) (1985)
`1023–1029.
`testicular 17␣-hydroxy-
`[20] M. Ayub, M.J. Levell, Inhibition of rat
`lase
`and 17,20-lyase
`activities by anti-androgens
`(flutamide,
`hydroxyflutamide, RU23908, cyproterone acetate) in vitro, J. Steroid
`Biochem. 28 (1) (1987) 43–47.
`[21] M.M. Bradford, A rapid and sensitive method for the quantitation
`of ␮g quantities of protein utilizing the principle of protein dye
`binding, Anal. Biochem. 72 (1976) 248–254.
`[22] S.E. Barrie, M.G. Rowlands, A.B. Foster, M. Jarman, Inhibition of
`17␣-hydroxylase/C17-C20 lyase by bifluranol and its analogues, J.
`Steroid Biochem. 33 (6) (1989) 1191–1195.
`[23] M.E. Lombardo, S.I. Hakky, M.K. Hall, P.B. Hudson, A study
`of androgen biosynthesis by the human testis in vitro, J. Steroid
`Biochem. Mol. Biol. 44 (2) (1993) 191–198.
`[24] G.T. Klus, J. Nakamura, J.-S. Li, Y.-Z. Ling, C. Son, J.A.
`Kemppainen, E.M. Wilson, A.M.H. Brodie, Growth inhibition of
`human prostate cells in vitro by novel
`inhibitors of androgen
`synthesis, Cancer Res. 56 (1996) 4956–4964.
`[25] J.-S. Li, Y. Li, C. Son, A.M.H. Brodie, Synthesis and evaluation
`of pregnane derivatives as inhibitors of human testicular 17␣-
`hydroxylase/C17,20-lyase, J. Med. Chem. 39 (1996) 4335–4339.
`[26] S.L. Miller, J.N. Wright, D.L. Corina, M. Akhtar, Mechanistic
`studies on pregnene side-chain cleavage enzyme (17␣-hydroxylase-
`
`

`

`542
`
`I. Duc et al. / Journal of Steroid Biochemistry & Molecular Biology 84 (2003) 537–542
`
`17,20-lyase) using 18O, J. Chem. Soc. Chem. Commun. (1991)
`157–159.
`[27] M. Akhtar, V.C.O. Njar, J.N. Wright, Mechanistic studies on
`aromatase and related C–C bond cleaving P-450 enzymes, J. Steroid
`Biochem. Mol. Biol. 44 (4–6) (1993) 375–387.
`[28] V.C.O. Njar, G.T. Klus, H.H. Johnson, A.M.H. Brodie, Synthesis of
`novel 21-trifluoropregnane steroids: inhibitors of 17␣-hydroxylase/
`17,20-lyase (17␣-lyase), Steroids 62 (1997) 468–473.
`
`[29] S.E. Barrie, G.A. Potter, M. Jarman, M. Dowsett, Highly potent
`inhibitors of human cytochrome P-450(17␣): activity in vitro and in
`vivo, Br. J. Cancer. 67 (Suppl.) (1993) 75.
`[30] S.E. Barrie, G.A. Potter, P.M. Goddard, B.P. Haynes, M. Dowsett, M.
`Jarman, Pharmacology of novel steroidal inhibitors of cytochrome
`P45017␣ (17␣-hydroxylase/C17-20 lyase), J. Steroid Biochem. Mol.
`Biol. 50 (5/6) (1994) 267–273.
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket